Vascular Cell
OPEN ACCESS  |  Review
Author Popup

Nanotechnology-mediated targeting of tumor angiogenesis

  • Deboshri Banerjee 1
  • Rania Harfouche 1
  • Shiladitya Sengupta 1 c @

@ corresponding author,  & equal contributor

Vascular Cell. 2011; 3(1):3 | © Banerjee et al

Received: 23 August  2010 | Accepted: 31 January  2011 | Published: 31 January  2011

Vascular Cell ISSN: 2045-824X

DOI: https://doi.org/10.1186/2045-824X-3-3

Author information Copyright & License
  • 1BWH-HST Center for Biomedical Engineering, Department of Medicine, Harvard-MIT Division of Health Science and Technology - Brigham and Women's Hospital, Harvard Medical SchoolCambridge, MA 02139, USA

Abstract


Angiogenesis is disregulated in many diseased states, most notably in cancer. An emerging strategy for the development of therapies targeting tumor-associated angiogenesis is to harness the potential of nanotechnology to improve the pharmacology of chemotherapeutics, including anti-angiogenic agents. Nanoparticles confer several advantages over that of free drugs, including their capability to carry high payloads of therapeutic agents, confer increased half-life and reduced toxicity to the drugs, and provide means for selective targeting of the tumor tissue and vasculature. The plethora of nanovectors available, in addition to the various methods available to combine them with anti-angiogenic drugs, allows researchers to fine-tune the pharmacological profile of the drugs ad infinitum. Use of nanovectors has also opened up novel avenues for non-invasive imaging of tumor angiogenesis. Herein, we review the types of nanovector and therapeutic/diagnostic agent combinations used in targeting tumor angiogenesis.

Introduction


Since Judah Folkman emphasized the 'angiogenic switch' hypothesis for tumor progression in 1991, there has been a tremendous surge in targeting angiogenesis for cancer therapeutics [1]. In the past 30 years, many advances have been made in the field, with the elucidation of various angiogenic molecules that could be targeted to halt angiogenesis, and hence, tumor progression. Angiogenesis, the formation of new capillaries from preexisting vessels, is crucial for ensuring normal embryonic vascular development of all vertebrates, as well as regulating physiological processes such as menses and wound healing in adults [24]. Deregulation of angiogenesis hence underlies pathologies characterized by vessel overgrowth (e.g. cancer) as well as vessel insufficiency (e.g. cardiovascular disease, CVD) [4].

It is now well-established that without angiogenesis, tumors cannot grow more than 2 mm in diameter [57]. Studies in breast cancer patients have showed that angiogenesis positively correlates with the degree of metastasis, tumor recurrence and shorter survival rates, thus demonstrating the value of angiogenesis as a prognostic cancer marker [1, 8]. Tumor angiogenesis essentially entails the same sequences of events as physiological angiogenesis, however, the latter proceeds in an uncontrolled and excessive manner giving rise to leaky and tortuous vessels that are in a constant state of inflammation [6, 9]. This is mainly due by an upregulation of angiogenic cytokines and growth factors, most notably the vascular endothelial cell growth factor (VEGF) and Angiopoietin (Ang) families, as well as integrins [1012]. Integrin αvβ3 is the best-characterized heterodimer that is upregulated in most cancer settings, both on the vasculature and on the tumor cells themselves [13, 14]. It is hence not surprising that these molecules are often targeted in both experimental and clinical cancer settings.

As such, the first U.S. Food and Drug Administration (FDA) approved anti-angiogenic therapy was the monoclonal antibody Bevacizumab (Avastin), that targets VEGF proteins overexpressed on colorectal cancer cells and their vasculature [15, 16]. In spite of the clinical success of Avastin, the majority of other such anti-angiogenic therapeutic agents have yet to pass phase II clinical trials, suggesting a new paradigm is essential to target aberrant angiogenesis.

Moving away from conventional chemotherapy

Engineering anti-angiogenic nanoparticles to suit our needs: Playing with nanovector backbone and drug coupling for therapeutic and imaging purposes

Since nanoparticles were first proposed by Marty JJ. et al. in 1978 as novel drug-delivery systems [39], their use as anti-cancer agents exploded during the 1980 s. However, only more recently (1995) have they been used to target angiogenesis [40]. Several nanovectors have been reported thus far in mediating anti-angiogenesis therapy and imaging of the tumor vasculature. These include an arsenal of synthetic and natural nanoparticles such as polymeric conjugates and polymeric nanoparticles; liposomes and micelles; synthetic organic nanoparticles such as dendrimers; carbon-based nanostructures such as carbon nanotubes and polyhydroxylated fullerenes; inorganic nanoparticles of gold, silver and iron-oxide; quantum dots; viral capsids and ferritin. The plethora of nanovectors allows researchers to fine-tune the properties of the drugs depending on their target. Further fine-tuning is also possible depending on the method of drug-nanovector coupling, thus offering the potential to engineer revolutionary therapeutics in the field of angiogenesis. Herein, we review the different types of nanovectors that have been studied to formulate anti-angiogenic agents for imaging and therapeutic purposes, their main modifications, as well as their advantages and limitations.

New generation research in anti-angiogenesis therapy

Apart from the more conventional approaches of arraying small molecule chemotherapeutic drugs or antibodies on different synthetic or natural nanovectors to achieve anti-angiogenic effects, new research reports are emerging that target the molecular mechanism of angiogenesis by using approaches such as gene silencing and others. In the following sections, we will review some of these emerging new strategies.

Concluding Remarks and Future Directions


The tumor neovasculature is an attractive target for anti-angiogenic therapy as well as non-invasive imaging studies. Nanotechnology has emerged as an exciting field in this area of research due to multiple advantages, including the capacity of nanoparticles to carry multiple moities of therapeutic and imaging agents, offer longer circulation time and increase the therapeutic index of chemotherapeutcs, to name a few. Moreover, with the various types of nanovectors available, many of which are FDA-approved, along with the various methods for coupling them to drugs and diagnostic agents, there is an endless opportunity to fine-tune nanotherapeutics depending on the task needed. Clearly, the advent of nanothechnology provides a huge potential for devising increasingly novel anti-angiogenic therapeutics that can eventually be translated from bench to bed-side.

Acknowledgements


The authors would like to thank Dr. Richard M. White and Dr. Leonard I. Zon (Children's Hospital Boston) for providing Casper Zebrafish and for their advice, as well as Dr. Dirk M. Hentschel and Dr. Joseph V. Bonventre (Harvard Institutes of Medicine, Boston, MA) for access to Zebrafish facilities. This work was funded by a CIHR fellowship to Rania Harfouche, and a Department of Defense BCRP Era of Hope Scholar Award (W81XWH-07-1-0482) and a BCRP Innovator Collaborative Award, and a NIH RO1 (1R01CA135242-01A2) to Shiladitya Sengupta.

Authors’ original submitted files for images


Below are the links to the authors’ original submitted files for images.

Authors’ original file for figure 1

Click here to view.

Authors’ original file for figure 2

Click here to view.

Authors’ original file for figure 3

Click here to view.

Authors’ original file for figure 4

Click here to view.

Authors’ original file for figure 5

Click here to view.

References

  1. Weidner N, Semple JP, Welch WR, Folkman J.  Tumor angiogenesis and metastasis-- correlation in invasive breast carcinoma. N Engl J Med. 1991;324:1-8.
  2. Risau W.  Mechanisms of angiogenesis. Nature. 1997;386:671-674.
  3. Folkman J.  Angiogenesis in cancer, vascular, rheumatoid and other disease. Nat Med. 1995;1:27-31.
  4. Folkman J.  Tumor angiogenesis: therapeutic implications. N Engl J Med. 1971;285:1182-1186.
  5. Naumov GN, Akslen LA, Folkman J.  Role of angiogenesis in human tumor dormancy: animal models of the angiogenic switch. Cell Cycle. 2006;5:1779-1787.
  6. Chaplain MA.  Mathematical modelling of angiogenesis. J Neurooncol. 2000;50:37-51.
  7. Folkman J.  Incipient angiogenesis 1. J Natl Cancer Inst. 2000;92:94-95.
  8. Weidner N, Folkman J, Pozza F, Bevilacqua P, Allred EN, Moore DH, Meli S, Gasparini G.  Tumor angiogenesis: a new significant and independent prognostic indicator in early-stage breast carcinoma 1. J Natl Cancer Inst. 1992;84:1875-1887.
  9. Fukumura D, Jain RK.  Imaging angiogenesis and the microenvironment. APMIS. 2008;116:695-715.
  10. Boudreau N, Myers C.  Breast cancer-induced angiogenesis: multiple mechanisms and the role of the microenvironment. Breast Cancer Res. 2003;5:140-146.
  11. Khodarev NN, Yu J, Labay E, Darga T, Brown CK, Mauceri HJ, Yassari R, Gupta N, Weichselbaum RR.  Tumour-endothelium interactions in co-culture: coordinated changes of gene expression profiles and phenotypic properties of endothelial cells. J Cell Sci. 2003;116:1013-1022.
  12. Desgrosellier JS, Cheresh DA.  Integrins in cancer: biological implications and therapeutic opportunities. Nat Rev Cancer. 2010;10:9-22.
  13. Yeh CH, Peng HC, Huang TF.  Accutin, a new disintegrin, inhibits angiogenesis in vitro and in vivo by acting as integrin alphavbeta3 antagonist and inducing apoptosis. Blood. 1998;92:3268-3276.
  14. Liekens S, De Clercq E, Neyts J.  Angiogenesis: regulators and clinical applications. Biochem Pharmacol. 2001;61:253-270.
  15. Peer D, Karp JM, Hong S, Farokhzad OC, Margalit R, Langer R.  Nanocarriers as an emerging platform for cancer therapy. Nat Nanotechnol. 2007;2:751-760.
  16. Ferrara N.  VEGF as a therapeutic target in cancer. Oncology. 2005;69(Suppl 3):11-16.
  17. Folkman J.  Fundamental concepts of the angiogenic process. Curr Mol Med. 2003;3:643-651.
  18. Segal E, Satchi-Fainaro R.  Design and development of polymer conjugates as anti-angiogenic agents. Adv Drug Deliv Rev. 2009;61:1159-1176.
  19. Kerbel RS, Kamen BA.  The anti-angiogenic basis of metronomic chemotherapy. Nat Rev Cancer. 2004;4:423-436.
  20. Davis ME, Chen ZG, Shin DM.  Nanoparticle therapeutics: an emerging treatment modality for cancer. Nat Rev Drug Discov. 2008;7:771-782.
  21. Suri SS, Fenniri H, Singh B.  Nanotechnology-based drug delivery systems. J Occup Med Toxicol. 2007;2:16-.
  22. Farokhzad OC, Langer R.  Impact of nanotechnology on drug delivery. ACS Nano. 2009;3:16-20.
  23. Couvreur P, Vauthier C.  Nanotechnology: intelligent design to treat complex disease. Pharm Res. 2006;23:1417-1450.
  24. Satchi-Fainaro R, Mamluk R, Wang L, Short SM, Nagy JA, Feng D, Dvorak AM, Dvorak HF, Puder M, Mukhopadhyay D, Folkman J.  Inhibition of vessel permeability by TNP-470 and its polymer conjugate, caplostatin. Cancer Cell. 2005;7:251-261.
  25. Satchi-Fainaro R, Puder M, Davies JW, Tran HT, Sampson DA, Greene AK, Corfas G, Folkman J.  Targeting angiogenesis with a conjugate of HPMA copolymer and TNP-470. Nat Med. 2004;10:255-261.
  26. Sengupta S, Eavarone D, Capila I, Zhao G, Watson N, Kiziltepe T, Sasisekharan R.  Temporal targeting of tumour cells and neovasculature with a nanoscale delivery system. Nature. 2005;436:568-572.
  27. Chaudhuri P, Harfouche R, Soni S, Hentschel DM, Sengupta S.  Shape effect of carbon nanovectors on angiogenesis. ACS Nano. 2010;4:574-582.
  28. Harfouche R, Basu S, Soni S, Hentschel DM, Mashelkar RA, Sengupta S.  Nanoparticle-mediated targeting of phosphatidylinositol-3-kinase signaling inhibits angiogenesis. Angiogenesis. 2009.
  29. Anderson SA, Rader RK, Westlin WF, Null C, Jackson D, Lanza GM, Wickline SA, Kotyk JJ.  Magnetic resonance contrast enhancement of neovasculature with alpha(v)beta(3)-targeted nanoparticles. Magn Reson Med. 2000;44:433-439.
  30. Park JH, Kwon S, Nam JO, Park RW, Chung H, Seo SB, Kim IS, Kwon IC, Jeong SY.  Self-assembled nanoparticles based on glycol chitosan bearing 5beta-cholanic acid for RGD peptide delivery. J Control Release. 2004;95:579-588.
  31. Winter PM, Morawski AM, Caruthers SD, Fuhrhop RW, Zhang H, Williams TA, Allen JS, Lacy EK, Robertson JD, Lanza GM, Wickline SA.  Molecular imaging of angiogenesis in early-stage atherosclerosis with alpha(v)beta3-integrin-targeted nanoparticles. Circulation. 2003;108:2270-2274.
  32. Waters EA, Chen J, Yang X, Zhang H, Neumann R, Santeford A, Arbeit J, Lanza GM, Wickline SA.  Detection of targeted perfluorocarbon nanoparticle binding using 19F diffusion weighted MR spectroscopy. Magn Reson Med. 2008;60:1232-1236.
  33. Rong J, Habuchi H, Kimata K, Lindahl U, Kusche-Gullberg M.  Substrate specificity of the heparan sulfate hexuronic acid 2-O-sulfotransferase. Biochemistry. 2001;40:5548-5555.
  34. Xie J, Shen Z, Li KC, Danthi N.  Tumor angiogenic endothelial cell targeting by a novel integrin-targeted nanoparticle. Int J Nanomedicine. 2007;2:479-485.
  35. Buehler A, van Zandvoort MA, Stelt BJ, Hackeng TM, Schrans-Stassen BH, Bennaghmouch A, Hofstra L, Cleutjens JP, Duijvestijn A, Smeets MB, de Kleijn DP, Post MJ, de Muinck ED.  cNGR: a novel homing sequence for CD13/APN targeted molecular imaging of murine cardiac angiogenesis in vivo. Arterioscler Thromb Vasc Biol. 2006;26:2681-2687.
  36. Hebbar M, Peyrat JP.  Significance of soluble endothelial molecule E-selectin in patients with breast cancer. Int J Biol Markers. 2000;15:15-21.
  37. Kang HW, Josephson L, Petrovsky A, Weissleder R, Bogdanov A.  Magnetic resonance imaging of inducible E-selectin expression in human endothelial cell culture. Bioconjug Chem. 2002;13:122-127.
  38. Wagner S, Rothweiler F, Anhorn MG, Sauer D, Riemann I, Weiss EC, Katsen-Globa A, Michaelis M, Cinatl J, Schwartz D, Kreuter J, von BH, Langer K.  Enhanced drug targeting by attachment of an anti alphav integrin antibody to doxorubicin loaded human serum albumin nanoparticles. Biomaterials. 2010;31:2388-2398.
  39. Marty JJ, Oppenheim RC, Speiser P.  Nanoparticles--a new colloidal drug delivery system. Pharm Acta Helv. 1978;53:17-23.
  40. Winet H, Hollinger JO, Stevanovic M.  Incorporation of polylactide-polyglycolide in a cortical defect: neoangiogenesis and blood supply in a bone chamber. J Orthop Res. 1995;13:679-689.
  41. Couvreur P, Kante B, Roland M, Speiser P.  Adsorption of antineoplastic drugs to polyalkylcyanoacrylate nanoparticles and their release in calf serum. J Pharm Sci. 1979;68:1521-1524.
  42. Segal E, Pan H, Ofek P, Udagawa T, Kopeckova P, Kopecek J, Satchi-Fainaro R.  Targeting angiogenesis-dependent calcified neoplasms using combined polymer therapeutics. PLoS ONE. 2009;4:e5233-.
  43. Miller K, Erez R, Segal E, Shabat D, Satchi-Fainaro R.  Targeting bone metastases with a bispecific anticancer and antiangiogenic polymer-alendronate-taxane conjugate. Angew Chem Int Ed Engl. 2009;48:2949-2954.
  44. Mitra A, Mulholland J, Nan A, McNeill E, Ghandehari H, Line BR.  Targeting tumor angiogenic vasculature using polymer-RGD conjugates. J Control Release. 2005;102:191-201.
  45. Mitra A, Coleman T, Borgman M, Nan A, Ghandehari H, Line BR.  Polymeric conjugates of mono- and bi-cyclic alphaVbeta3 binding peptides for tumor targeting. J Control Release. 2006;114:175-183.
  46. Basu S, Harfouche R, Soni S, Chimote G, Mashelkar RA, Sengupta S.  Nanoparticle-mediated targeting of MAPK signaling predisposes tumor to chemotherapy. Proc Natl Acad Sci USA. 2009;106:7957-7961.
  47. Veronese FM, Pasut G.  PEGylation, successful approach to drug delivery. Drug Discov Today. 2005;10:1451-1458.
  48. White RM, Sessa A, Burke C, Bowman T, LeBlanc J, Ceol C, Bourque C, Dovey M, Goessling W, Burns CE, Zon LI.  Transparent adult zebrafish as a tool for in vivo transplantation analysis. Cell Stem Cell. 2008;2:183-189.
  49. Anand P, Nair HB, Sung B, Kunnumakkara AB, Yadav VR, Tekmal RR, Aggarwal BB.  Design of curcumin-loaded PLGA nanoparticles formulation with enhanced cellular uptake, and increased bioactivity in vitro and superior bioavailability in vivo. Biochem Pharmacol. 2010;79:330-338.
  50. Gillies ER, Frechet JM.  Dendrimers and dendritic polymers in drug delivery. Drug Discov Today. 2005;10:35-43.
  51. Xu Y, Wen Z, Xu Z.  Chitosan nanoparticles inhibit the growth of human hepatocellular carcinoma xenografts through an antiangiogenic mechanism. Anticancer Res. 2009;29:5103-5109.
  52. Backer MV, Gaynutdinov TI, Patel V, Bandyopadhyaya AK, Thirumamagal BT, Tjarks W, Barth RF, Claffey K, Backer JM.  Vascular endothelial growth factor selectively targets boronated dendrimers to tumor vasculature. Mol Cancer Ther. 2005;4:1423-1429.
  53. Benny O, Fainaru O, Adini A, Cassiola F, Bazinet L, Adini I, Pravda E, Nahmias Y, Koirala S, Corfas G, D'Amato RJ, Folkman J.  An orally delivered small-molecule formulation with antiangiogenic and anticancer activity. Nat Biotechnol. 2008;26:799-807.
  54. Nasongkla N, Shuai X, Ai H, Weinberg BD, Pink J, Boothman DA, Gao J.  cRGD-functionalized polymer micelles for targeted doxorubicin delivery. Angew Chem Int Ed Engl. 2004;43:6323-6327.
  55. Oku N, Asai T, Watanabe K, Kuromi K, Nagatsuka M, Kurohane K, Kikkawa H, Ogino K, Tanaka M, Ishikawa D, Tsukada H, Momose M, Nakayama J, Taki T.  Anti-neovascular therapy using novel peptides homing to angiogenic vessels. Oncogene. 2002;21:2662-2669.
  56. Asai T, Shimizu K, Kondo M, Kuromi K, Watanabe K, Ogino K, Taki T, Shuto S, Matsuda A, Oku N.  Anti-neovascular therapy by liposomal DPP-CNDAC targeted to angiogenic vessels. FEBS Lett. 2002;520:167-170.
  57. Maeda N, Takeuchi Y, Takada M, Sadzuka Y, Namba Y, Oku N.  Anti-neovascular therapy by use of tumor neovasculature-targeted long-circulating liposome. J Control Release. 2004;100:41-52.
  58. Shimizu K, Asai T, Oku N.  Antineovascular therapy, a novel antiangiogenic approach. Expert Opin Ther Targets. 2005;9:63-76.
  59. Li L, Wartchow CA, Danthi SN, Shen Z, Dechene N, Pease J, Choi HS, Doede T, Chu P, Ning S, Lee DY, Bednarski MD, Knox SJ.  A novel antiangiogenesis therapy using an integrin antagonist or anti-Flk-1 antibody coated 90Y-labeled nanoparticles. Int J Radiat Oncol Biol Phys. 2004;58:1215-1227.
  60. Hatakeyama H, Akita H, Ishida E, Hashimoto K, Kobayashi H, Aoki T, Yasuda J, Obata K, Kikuchi H, Ishida T, Kiwada H, Harashima H.  Tumor targeting of doxorubicin by anti-MT1-MMP antibody-modified PEG liposomes. Int J Pharm. 2007;342:194-200.
  61. Murugesan S, Mousa SA, O'connor LJ, Lincoln DW, Linhardt RJ.  Carbon inhibits vascular endothelial growth factor-and fibroblast growth factor-promoted angiogenesis. FEBS Lett. 2007;581:1157-1160.
  62. Mukherjee P, Bhattacharya R, Wang P, Wang L, Basu S, Nagy JA, Atala A, Mukhopadhyay D, Soker S.  Antiangiogenic properties of gold nanoparticles. Clin Cancer Res. 2005;11:3530-3534.
  63. Gurunathan S, Lee KJ, Kalishwaralal K, Sheikpranbabu S, Vaidyanathan R, Eom SH.  Antiangiogenic properties of silver nanoparticles. Biomaterials. 2009;30:6341-6350.
  64. McCarthy JR, Kelly KA, Sun EY, Weissleder R.  Targeted delivery of multifunctional magnetic nanoparticles. Nanomedicine (Lond). 2007;2:153-167.
  65. Chen J, Wu H, Han D, Xie C.  Using anti-VEGF McAb and magnetic nanoparticles as double-targeting vector for the radioimmunotherapy of liver cancer. Cancer Lett. 2006;231:169-175.
  66. Maeng JH, Lee DH, Jung KH, Bae YH, Park IS, Jeong S, Jeon YS, Shim CK, Kim W, Kim J, Lee J, Lee YM, Kim JH, Kim WH, Hong SS.  Multifunctional doxorubicin loaded superparamagnetic iron oxide nanoparticles for chemotherapy and magnetic resonance imaging in liver cancer. Biomaterials. 2010;31:4995-5006.
  67. Cristofanilli M, Charnsangavej C, Hortobagyi GN.  Angiogenesis modulation in cancer research: novel clinical approaches. Nat Rev Drug Discov. 2002;1:415-426.
  68. Cormode DP, Jarzyna PA, Mulder WJ, Fayad ZA.  Modified natural nanoparticles as contrast agents for medical imaging. Adv Drug Deliv Rev. 2010;62:329-338.
  69. Drevs J, Muller-Driver R, Wittig C, Fuxius S, Esser N, Hugenschmidt H, Konerding MA, Allegrini PR, Wood J, Hennig J, Unger C, Marme D.  PTK787/ZK 222584, a specific vascular endothelial growth factor-receptor tyrosine kinase inhibitor, affects the anatomy of the tumor vascular bed and the functional vascular properties as detected by dynamic enhanced magnetic resonance imaging. Cancer Res. 2002;62:4015-4022.
  70. Reichardt W, Hu-Lowe D, Torres D, Weissleder R, Bogdanov A.  Imaging of VEGF receptor kinase inhibitor-induced antiangiogenic effects in drug-resistant human adenocarcinoma model. Neoplasia. 2005;7:847-853.
  71. Sipkins DA, Cheresh DA, Kazemi MR, Nevin LM, Bednarski MD, Li KC.  Detection of tumor angiogenesis in vivo by alphaVbeta3-targeted magnetic resonance imaging. Nat Med. 1998;4:623-626.
  72. Mulder WJ, Strijkers GJ, Habets JW, Bleeker EJ, van der Schaft DW, Storm G, Koning GA, Griffioen AW, Nicolay K.  MR molecular imaging and fluorescence microscopy for identification of activated tumor endothelium using a bimodal lipidic nanoparticle. FASEB J. 2005;19:2008-2010.
  73. Mulder WJ, van der Schaft DW, Hautvast PA, Strijkers GJ, Koning GA, Storm G, Mayo KH, Griffioen AW, Nicolay K.  Early in vivo assessment of angiostatic therapy efficacy by molecular MRI. FASEB J. 2007;21:378-383.
  74. Kluza E, van der Schaft DW, Hautvast PA, Mulder WJ, Mayo KH, Griffioen AW, Strijkers GJ, Nicolay K.  Synergistic targeting of alphavbeta3 integrin and galectin-1 with heteromultivalent paramagnetic liposomes for combined MR imaging and treatment of angiogenesis. Nano Lett. 2010;10:52-58.
  75. Samei E, Saunders RS, Badea CT, Ghaghada KB, Hedlund LW, Qi Y, Yuan H, Bentley RC, Mukundan S.  Micro-CT imaging of breast tumors in rodents using a liposomal, nanoparticle contrast agent. Int J Nanomedicine. 2009;4:277-282.
  76. Mulder WJ, Koole R, Brandwijk RJ, Storm G, Chin PT, Strijkers GJ, de Mello DC, Nicolay K, Griffioen AW.  Quantum dots with a paramagnetic coating as a bimodal molecular imaging probe. Nano Lett. 2006;6:1-6.
  77. Oostendorp M, Douma K, Hackeng TM, Dirksen A, Post MJ, van Zandvoort MA, Backes WH.  Quantitative molecular magnetic resonance imaging of tumor angiogenesis using cNGR-labeled paramagnetic quantum dots. Cancer Res. 2008;68:7676-7683.
  78. Manchester M, Singh P.  Virus-based nanoparticles (VNPs): platform technologies for diagnostic imaging. Adv Drug Deliv Rev. 2006;58:1505-1522.
  79. Banerjee D, Liu AP, Voss NR, Schmid SL, Finn MG.  Multivalent display and receptor-mediated endocytosis of transferrin on virus-like particles. Chembiochem. 2010;11:1273-1279.
  80. Lewis JD, Destito G, Zijlstra A, Gonzalez MJ, Quigley JP, Manchester M, Stuhlmann H.  Viral nanoparticles as tools for intravital vascular imaging. Nat Med. 2006;12:354-360.
  81. Brunel FM, Lewis JD, Destito G, Steinmetz NF, Manchester M, Stuhlmann H, Dawson PE.  Hydrazone ligation strategy to assemble multifunctional viral nanoparticles for cell imaging and tumor targeting. Nano Lett. 2010;10:1093-1097.
  82. Geninatti CS, Bussolati B, Tei L, Grange C, Esposito G, Lanzardo S, Camussi G, Aime S.  Magnetic resonance visualization of tumor angiogenesis by targeting neural cell adhesion molecules with the highly sensitive gadolinium-loaded apoferritin probe. Cancer Res. 2006;66:9196-9201.
  83. Kim WJ, Yockman JW, Lee M, Jeong JH, Kim YH, Kim SW.  Soluble Flt-1 gene delivery using PEI-g-PEG-RGD conjugate for anti-angiogenesis. J Control Release. 2005;106:224-234.
  84. Kim WJ, Yockman JW, Jeong JH, Christensen LV, Lee M, Kim YH, Kim SW.  Anti-angiogenic inhibition of tumor growth by systemic delivery of PEI-g-PEG-RGD/pCMV-sFlt-1 complexes in tumor-bearing mice. J Control Release. 2006;114:381-388.
  85. Hadj-Slimane R, Lepelletier Y, Lopez N, Garbay C, Raynaud F.  Short interfering RNA (siRNA), a novel therapeutic tool acting on angiogenesis. Biochimie. 2007;89:1234-1244.
  86. Schiffelers RM, Ansari A, Xu J, Zhou Q, Tang Q, Storm G, Molema G, Lu PY, Scaria PV, Woodle MC.  Cancer siRNA therapy by tumor selective delivery with ligand-targeted sterically stabilized nanoparticle. Nucleic Acids Res. 2004;32:e149-.
  87. Villares GJ, Zigler M, Wang H, Melnikova VO, Wu H, Friedman R, Leslie MC, Vivas-Mejia PE, Lopez-Berestein G, Sood AK, Bar-Eli M.  Targeting melanoma growth and metastasis with systemic delivery of liposome-incorporated protease-activated receptor-1 small interfering RNA. Cancer Res. 2008;68:9078-9086.
  88. Pille JY, Li H, Blot E, Bertrand JR, Pritchard LL, Opolon P, Maksimenko A, Lu H, Vannier JP, Soria J, Malvy C, Soria C.  Intravenous delivery of anti-RhoA small interfering RNA loaded in nanoparticles of chitosan in mice: safety and efficacy in xenografted aggressive breast cancer. Hum Gene Ther. 2006;17:1019-1026.
  89. Khati M.  The future of aptamers in medicine. J Clin Pathol. 2010.
  90. Lee JH, Yigit MV, Mazumdar D, Lu Y.  Molecular diagnostic and drug delivery agents based on aptamer-nanomaterial conjugates. Adv Drug Deliv Rev. 2010.
  91. Crawford M, Woodman R, Ko FP.  Peptide aptamers: tools for biology and drug discovery. Brief Funct Genomic Proteomic. 2003;2:72-79.
  92. Wang W, Chen C, Qian M, Zhao XS.  Aptamer biosensor for protein detection using gold nanoparticles. Anal Biochem. 2008;373:213-219.
  93. Estevez MC, Huang YF, Kang H, O'Donoghue MB, Bamrungsap S, Yan J, Chen X, Tan W.  Nanoparticle-aptamer conjugates for cancer cell targeting and detection. Methods Mol Biol. 2010;624:235-248.
  94. Smith JE, Medley CD, Tang Z, Shangguan D, Lofton C, Tan W.  Aptamer-conjugated nanoparticles for the collection and detection of multiple cancer cells. Anal Chem. 2007;79:3075-3082.
  95. Carrasquillo KG, Ricker JA, Rigas IK, Miller JW, Gragoudas ES, Adamis AP.  Controlled delivery of the anti-VEGF aptamer EYE001 with poly(lactic-co-glycolic)acid microspheres. Invest Ophthalmol Vis Sci. 2003;44:290-299.
  96. Willis MC, Collins BD, Zhang T, Green LS, Sebesta DP, Bell C, Kellogg E, Gill SC, Magallanez A, Knauer S, Bendele RA, Gill PS, Janjic N.  Liposome-anchored vascular endothelial growth factor aptamers. Bioconjug Chem. 1998;9:573-582.
  97. White RR, Shan S, Rusconi CP, Shetty G, Dewhirst MW, Kontos CD, Sullenger BA.  Inhibition of rat corneal angiogenesis by a nuclease-resistant RNA aptamer specific for angiopoietin-2. Proc Natl Acad Sci USA. 2003;100:5028-5033.
  98. Harfouche R, Hussain SN.  Signaling and regulation of endothelial cell survival by angiopoietin-2. Am J Physiol Heart Circ Physiol. 2006;291:H1635-H1645.
  99. Mern DS, Hoppe-Seyler K, Hoppe-Seyler F, Hasskarl J, Burwinkel B.  Targeting Id1 and Id3 by a specific peptide aptamer induces E-box promoter activity, cell cycle arrest, and apoptosis in breast cancer cells. Breast Cancer Res Treat. 2010.
  100. Zhao BM, Hoffmann FM.  Inhibition of transforming growth factor-beta1-induced signaling and epithelial-to-mesenchymal transition by the Smad-binding peptide aptamer Trx-SARA. Mol Biol Cell. 2006;17:3819-3831.
  101. Matsumura S, Sato S, Yudasaka M, Tomida A, Tsuruo T, Iijima S, Shiba K.  Prevention of carbon nanohorn agglomeration using a conjugate composed of comb-shaped polyethylene glycol and a peptide aptamer. Mol Pharm. 2009;6:441-447.
  102. Anderson SA, Glod J, Arbab AS, Noel M, Ashari P, Fine HA, Frank JA.  Noninvasive MR imaging of magnetically labeled stem cells to directly identify neovasculature in a glioma model. Blood. 2005;105:420-425.
  103. Yang F, Cho SW, Son SM, Bogatyrev SR, Singh D, Green JJ, Mei Y, Park S, Bhang SH, Kim BS, Langer R, Anderson DG.  Genetic engineering of human stem cells for enhanced angiogenesis using biodegradable polymeric nanoparticles. Proc Natl Acad Sci USA. 2010;107:3317-3322.
Publiverse Online Logo

The present article has been published in Vascular Cell journal by Publiverse Online S.R.L.